Language selection

Search

Patent 3070303 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3070303
(54) English Title: PLASMINOGEN ACTIVATOR INHIBITOR-1 (PAI-1) INHIBITOR AND METHOD OF USE
(54) French Title: INHIBITEUR DE L'ACTIVATEUR DU PLASMINOGENE-1 (PAI-1) ET PROCEDE D'UTILISATION
Status: Examination Requested
Bibliographic Data
(51) International Patent Classification (IPC):
  • A01N 43/42 (2006.01)
  • A61K 31/47 (2006.01)
  • C12Q 1/68 (2018.01)
(72) Inventors :
  • LAWRENCE, DANIEL A. (United States of America)
  • EMAL, CORY (United States of America)
  • REINKE, ASHLEY (United States of America)
  • LI, SHIH-HON (United States of America)
(73) Owners :
  • THE REGENTS OF THE UNIVERSITY OF MICHIGAN (United States of America)
  • EASTERN MICHIGAN UNIVERSITY (United States of America)
The common representative is: THE REGENTS OF THE UNIVERSITY OF MICHIGAN
(71) Applicants :
  • THE REGENTS OF THE UNIVERSITY OF MICHIGAN (United States of America)
  • EASTERN MICHIGAN UNIVERSITY (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2018-07-27
(87) Open to Public Inspection: 2019-01-31
Examination requested: 2023-07-25
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2018/043998
(87) International Publication Number: WO2019/023526
(85) National Entry: 2020-01-16

(30) Application Priority Data:
Application No. Country/Territory Date
62/537,513 United States of America 2017-07-27

Abstracts

English Abstract


Provided herein are plasminogen activator-1 (PAI-1 ) inhibitor compounds and
uses thereof in the treatment of any
disease or disorder associated with elevated PAI-1. The disclosure includes,
but is not limited to, the use of such compounds to prevent
or reduce thrombosis and fibrosis, to promote thrombolysis, and to modulate
lipid metabolism and treat diseases or disorders associated
with elevated PAI-1, cholesterol, or lipid levels.


French Abstract

La présente invention concerne des composés inhibiteurs de l'activateur du plasminogène-1 (PAI-1) et leurs utilisations dans le traitement de toute maladie ou trouble associé(e) à des niveaux élevés de PAI-1. La présente invention concerne, mais n'est pas limitée, à l'utilisation de tels composés pour prévenir ou réduire la thrombose et la fibrose, pour favoriser la thrombolyse, et moduler le métabolisme des lipides et traiter les maladies ou les troubles associés/associées à des niveaux élevés de PAI-1, de cholestérol, ou de lipide.

Claims

Note: Claims are shown in the official language in which they were submitted.


What is Claimed:
1. A compound having a structure of:
Image
wherein X is F or Cl,
or a pharmaceutically acceptable salt thereof.
2. The compound or salt of claim 1 having a structure of:
Image
3. The compound or salt of claim 1 having a structure of:
Image
4. A compound having a
structure Image , or a
pharmaceutically acceptable salt thereof.
5. A pharmaceutical composition comprising the compound or salt of any one
of
claims 1 to 4 or a compound having a structure of Image
or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable excipient.
6. The pharmaceutical composition of claim 5 wherein the compound has a
structure of Image or a pharmaceutically acceptable salt thereof.
26

7. A method of inhibiting plasminogen activator inhibitor-1 (PAI-1)
comprising
contacting PAI-1 with the compound, salt, or composition of any one of claims
1 to 6 in an
amount effective the inhibit PAI-1.
8. A method of treating a disorder associated with aberrant PAI-1 activity,

comprising administering to a subject in need thereof the compound, salt, or
composition of
any one of claims 1 to 6 in an amount effective to treat the disorder.
9. The method of claim 8, wherein the disorder is cancer, septicemia,
obesity,
insulin resistance, a disease or disorder associated with dysregulation of
lipid metabolism, a
disease or disorder associated with an elevated level of VLDL or LDL, high
cholesterol, a
proliferative disease or disorder, fibrosis and fibrotic disease, inflammatory
bowel disease,
coagulation homeostasis, cerebrovascular disease, microvascular disease,
hypertension,
dementia, atherosclerosis, osteoporosis, osteopenia, arthritis, asthma, heart
failure,
arrhythmia, angina, hormone insufficiency, Alzheimer's disease, hypertension,
inflammation,
sepsis, fibrinolytic disorder, stroke, dementia, coronary heart disease,
myocardial infarction,
stable and unstable angina, vascular disease, peripheral arterial disease,
acute vascular
syndrome, thrombosis, prothrombosis, deep vein thrombosis, pulmonary embolism,

cerebrovascular disease, microvascular disease, hypertension, diabetes,
hyperglycemia,
hyperinsulinemia, malignant lesions, premalignant lesions, gastrointestinal
malignancies,
liposarcoma, epithelial tumor, and psoriasis, an extracellular matrix
accumulation disorder,
neoangiogenesis, myelofibrosis, fibrinolytic impairment, polycystic ovary
syndrome, bone
loss induced by estrogen deficiency, angiogenesis, neoangiogenesis,
myelofibrosis, or
fibrinolytic impairment.
10. The method of claim 9, wherein the disease or disorder involving
thrombosis
or prothrombosis is formation of atherosclerotic plaques, venous thrombosis,
arterial
thrombosis, myocardial ischemia, atrial fibrillation, deep vein thrombosis, a
coagulation
syndrome, pulmonary thrombosis, cerebral thrombosis, a thromboembolic
complication of
surgery, and peripheral arterial occlusion.
11. The method of claim 9, wherein the disorder is fibrosis.
12. The method of claim 11, wherein the fibrosis is pulmonary fibrosis,
renal
fibrosis, cardiac fibrosis, hepatic fibrosis, or scleroderma.
13. The method of claim 9, wherein the disorder is inflammatory bowel
disease.
14. The method of claim 13, wherein the inflammatory bowel disease is
Crohn's
disease or ulcerative colitis.
27

15. The method of claim 9, wherein the extracellular matrix accumulation
disorder
is renal fibrosis, chronic obstructive pulmonary disease, polycystic ovary
syndrome,
restenosis, renovascular disease, diabetic nephropathy, or organ transplant
rejection.
16. A method of modulating cholesterol, lipid clearance, and/or lipid
uptake in a
subject with an elevated level of PAI-1 comprising administering to the
subject an effective
amount of the compound, salt, or composition of any one of claims 1 to 6 in an
amount
effective to decrease the elevated level of PAI and modulate cholesterol,
lipid clearance,
and/or lipid uptake in the subject.
17. The method of claim 16, wherein the compound or salt increases
circulating
high density lipoprotein (HDL) and/or decreases circulating very low density
lipoprotein
(VLDL) in the subject.
18. The method of claim 16, wherein the compound or salt inhibits
apolipoprotein
E (ApoE) or apolipoprotein A (ApoA) binding to VLDL-R.
19. The method of claim 16, wherein the compound or salt decreases HDL or
apolipoprotein E (ApoE) or apolipoprotein A (ApoA) binding to an ApoA
receptor.
20. The method of claim 16, wherein the compound or salt decreases PAI-1
binding to apolipoprotein E (ApoE).
21. The method of claim 16, wherein the compound or salt decreases PAI-1
binding to apolipoprotein A (ApoA).
22. The method of claim 16, wherein the compound or salt decreases PAI-1
binding to VLDL.
23. The method of claim 16, wherein the compound or salt binds to PAI-1 in
the
presence of vitronectin.
24. The method of claim 16, wherein the compound or salt binds to PAI-1 in
the
presence of urokinase type plasminogen activator (uPA).
25. The method of any one of claims 7 to 24, wherein the subject is human.
28

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
PLASMINOGEN ACTIVATOR INHIBITOR-1 (PAI-1) INHIBITOR AND METHOD OF USE
STATEMENT OF GOVERNMENT SUPPORT
[0001] This invention was made with government support under HL089407 awarded
by
the National Institutes of Health. The government has certain rights in the
invention.
FIELD
[0002] Provided herein are compounds and methods for modulating plasminogen
activator
inhibitor-1 (PAI-1) activity. More particularly, the disclosure is directed to
inhibitors of PAI-1
and the uses of such inhibitors in regulating PAI-1 activity. Also provided
are uses of these
inhibitors for the treatment of many diseases or disorders associated with PAI-
1 activity.
Such diseases or disorders include, but are not limited to, dysregulation of
lipid metabolism,
obesity, diabetes, polycystic ovary syndrome, bone loss induced by estrogen
deficiency,
fibrosis and fibrotic disease, inflammation, cell migration and migration-
driven proliferation of
cells, angiogenesis, and thrombosis. Such inhibitors are also contemplated to
be useful for
modulation of endogenous fibrinolysis, and in conjunction with pharmacologic
thrombolysis.
BACKGROUND
[0003] Plasminogen activator inhibitor-1 (PAI-1) is a 50 kDa single-chain
glycoprotein that
is the principal inhibitor of both urokinase type plasminogen activator (uPA)
and tissue type
PA (tPA). PAI-1 inhibits tPA and uPA with second-order rate constants -107 M-
1s-1, a value
that is 10-1000 times faster than the rates of PA inhibition by other PAls.
Moreover,
approximately 70% of the total tPA in carefully collected normal human plasma
is detected in
complex with PAI-1, suggesting that inhibition of tPA by PAI-1 is a normal,
ongoing process.
PAI-1 can also directly inhibit plasmin. Thus, PAI-1 is the chief regulator of
plasmin
generation in vivo, and as such it appears to play an important role in both
fibrotic and
thrombotic disease. PAI-1 has three potential N-linked glycosylation sites and
contains
between 15 and 20% carbohydrate.
[0004] PAI-1 belongs to the Serine Protease Inhibitor super family (SERPIN),
which is a
gene family that includes many of the protease inhibitors found in blood, as
well as other
proteins with unrelated or unknown functions. Serpins are consumed in the
process of
protease inactivation and thus act as "suicide inhibitors." The association
between a serpin
and its target protease occurs at an amino acid residue, referred to as the
"bait" residue,
located on a surface loop of the serpin called the reactive center loop (RCL).
The "bait"
residue is also called the P1 residue, and is thought to mimic the normal
substrate of the
enzyme. Upon association of the P1 residue with the Si site of a target
protease, cleavage
of the RCL occurs. This is coupled to a large conformational change in the
serpin which
involves rapid insertion of the RCL into the major structural feature of a
serpin, 13-sheet A.
This results in tight docking of the protease to the serpin surface and to
distortion of the
1

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
enzyme structure, including its active site. RCL insertion also produces a
large increase in
serpin structural stability making the complex rigid and thus trapping the
protease in a
covalent acyl-enzyme complex with the serpin.
[0005] Native PAI-1 exists in at least two distinct conformations, an
active form that is
produced by cells and secreted, and an inactive or latent form that
accumulates in cell
culture medium over time. In blood and tissues, most of the PAI-1 is in the
active form;
however, in platelets both active and latent forms of PAI-1 are found. In
active PAI-1, the
RCL is exposed on the surface of the molecule, but upon reaction with a
protease, the
cleaved RCL integrates into the center of 6 sheet A. In the latent form, the
RCL is intact, but
instead of being exposed, the entire amino terminal side of the RCL is
inserted as the central
strand into the 6 sheet A. This accounts for the increased stability of latent
PAI-1 as well as
its lack of inhibitory activity.
[0006] Active PAI-1 spontaneously converts to the latent form with a half-life
of one to two
hours at 37 C, and latent PAI-1 can be converted back into the active form by
treatment with
denaturants. Negatively charged phospholipids can also convert latent PAI-1 to
the active
form, suggesting that cell surfaces may modulate PAI-1 activity. The
observation that latent
PAI-1 infused into rabbits is apparently converted to the active form is
consistent with this
hypothesis. The spontaneous reversible interconversion between the active and
latent
structures is unique for PAI-1 and distinguishes it from other serpins;
however, the biological
significance of the latent conformation remains unknown.
[0007] Other non-inhibitory forms of PAI-1 have also been identified. The
first form results
from oxidation of one or more critical methionine residues within active PAI-
1. This form
differs from latent PAI-1 in that it can be partially reactivated by an enzyme
that specifically
reduces oxidized methionine residues. Oxidative inactivation of PAI-1 may be
an additional
mechanism for the regulation of PAI-1, and oxygen radicals produced locally by
neutrophils
or other cells may inactivate PAI-1 and thus facilitate the generation of
plasmin at sites of
infection or in areas of tissue remodeling. PAI-1 also exists in two different
cleaved forms.
As noted above, PAI-1 in complex with a protease is cleaved in its RCL.
Uncomplexed PAI-
1 can also be found with its RCL cleaved, which can arise from dissociation of
PAI-1-PA
complexes or from cleavage of the RCL by a non-target protease at a site other
than the P1.
None of these forms of PAI-1 are able to inhibit protease activity; however,
they may interact
with other ligands.
[0008] The interaction of PAI-1 with non-protease ligands plays an essential
role in PAI-1
function. PAI-1 binds with high affinity to heparin, the cell adhesion protein
vitronectin, and
members the endocytic low-density lipoprotein receptor (LDL-R) family, such as
the
lipoprotein receptor-related protein (LRP), and the very low density
lipoprotein receptor
2

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
(VLDL-R). These non-protease interactions are important for both PAI-1
localization and
function, and they are largely conformationally controlled through structural
changes
associated with RCL insertion. In blood, most of the active PAI-1 circulates
in complex with
the glycoprotein vitronectin. The PAI-1 binding site for vitronectin has been
localized to a
region on the edge of p-sheet A in the PAI-1 structure. The binding site for
LDL-R family
members is less well characterized, but has been identified, in a region of
PAI-1 associated
with alpha helix D that is adjacent to the vitronectin binding domain. The
heparin binding
domain on PAI-1 has also been mapped. This site also localizes to alpha helix
D in a region
homologous to the heparin binding domain of antithrombin III, and may overlap
with the
binding site for LDL-R family members.
[0009] Vitronectin circulates in plasma and is present in the extracellular
matrix primarily
at sites of injury or remodeling. PAI-1 and vitronectin appear to have a
significant functional
interdependence. Vitronectin stabilizes PAI-1 in its active conformation,
thereby increasing
its biological half-life.
[0010] Vitronectin also enhances PAI-1 inhibitory efficiency for thrombin
approximately
300-fold. In turn, PAI-1 binding to vitronectin alters its conformation from
the native plasma
form, which does not support cell adhesion, to an "activated" form that is
competent to bind
integrins. However, integrin binding is blocked by the presence of PAI-1. As
noted above,
the association of PAI-1 with vitronectin is conformationally controlled and
upon inhibition of
a protease, the conformational change in PAI-1 associated with RCL insertion
results in a
loss of high affinity for vitronectin and a gain in affinity for LDL-R family
members. This is
due to RCL insertion in PAI-1, disrupting the vitronectin binding site, while
simultaneously
exposing a cryptic receptor binding site that is revealed only when PAI-1 is
in a complex with
a protease, which results in an approximately 100,000-fold shift in the
relative affinity of PAI-
1 from vitronectin to LDL-R family members and a subsequent shift in PAI-1
localization from
vitronectin to the cellular receptor. Thus, PAI-1 association with vitronectin
and LDL-R is
conformationally controlled.
[0011] High PAI-1 levels are associated with various diseases and disorders.
For
example, high PAI-1 levels are associated with acute diseases, such as sepsis
and
myocardial infarction, and chronic disorders, such as cancer, atherosclerosis,
and type 2
diabetes. In addition, high PAI-1 levels are associated with cardiovascular
disease, wherein
PAI-1 expression is significantly increased in severely atherosclerotic
vessels, and PAI-1
protein levels rise consistently during disease progression from normal
vessels to fatty
streaks to atherosclerotic plaques. Increased PAI-1 levels are also linked to
obesity, and
insulin resistance.
3

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0012] In addition, elevated plasma levels of PAI-1 have been associated with
thrombotic
events, and antibody neutralization of PAI-1 activity resulted in promotion of
endogenous
thrombolysis and reperfusion. Elevated levels of PAI-1 have also been
implicated in
polycystic ovary syndrome and bone loss induced by estrogen deficiency.
[0013] PAI-1 is synthesized in both murine and human adipocytes. There is also
a strong
correlation between the amount of visceral fat and plasma levels of PAI-1 in
humans and
mice. This dramatic up-regulation of PAI-1 in obesity has led to the
suggestion that adipose
tissue itself may directly contribute to elevated systemic PAI-1, which in-
turn increases the
probability of vascular disease through increased thrombosis, and accelerated
atherosclerosis. Notably, very recent data suggests that PAI-1 may also play a
direct role in
obesity.
[0014] In one study, genetically obese and diabetic ob/ob mice crossed into a
PAI-1
deficient background had significantly reduced body weight and improved
metabolic profiles
compared to ob/ob mice with PAI-1. Likewise, nutritionally-induced obesity and
insulin
resistance were dramatically attenuated in mice genetically deficient in PAI-1
and in mice
treated with an orally active PAI-1 inhibitor. The improved adiposity and
insulin resistance in
PAI-1-deficient mice may be related to the observation that PAI-1 deficient
mice on a high fat
diet had increased metabolic rates and total energy expenditure compared to
wild-type mice,
and peroxysome proliferator-activated receptor (PPARy) and adiponectin were
maintained.
However, the precise mechanism involved was not shown and may be complex,
since the
over-expression of PAI-1 in mice also impaired adipose tissue formation. Taken
together,
these observations suggest that PAI-1 plays a previously unrecognized direct
role in obesity
and insulin resistance that involves interactions beyond its identified
activities of modulating
fibrinolysis and tissue remodeling.
[0015] Indeed, if PAI-1 positively regulates adipose tissue development,
then the
association of increased PAI-1 expression with developing obesity may
constitute a positive
feedback loop promoting adipose tissue expansion and dysregulation of normal
cholesterol
homeostasis. Thus, there exists a need in the art for a greater understanding
of how PAI-1
is involved in metabolism, obesity and insulin resistance.
SUMMARY
0
H
Si N)..r N'NH2
H
0
x
[0016] Provided herein is a compound having a structure of OCF3 ,
wherein X is Cl or F, or a pharmaceutically acceptable salt thereof. In some
cases, the
4

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
0
H
SI N)YN,NH2
H
0
CI
compound is OCF3 , or a pharmaceutically acceptable salt
thereof. In
0
H
F0 N)YN,NH2
H
0
some cases, the compound is OCF3 , or a
pharmaceutically acceptable
0
H
40 F N)YN,NH2
H
0
salt thereof. Also provided is a PAI-1 inhibitor having a structure CI
or a pharmaceutically acceptable salt thereof. Further provided is a PAI-1
inhibitor having a
0
H
0 N)YN,NH2
H
0
CI
structure F , or a
pharmaceutically acceptable salt thereof. In
some cases, the compound is in the form of a pharmaceutically acceptable salt.
Further
provided are pharmaceutical compositions of one or more of the compounds or
salts
disclosed herein and a pharmaceutically acceptable excipient. In some cases,
the
0
H
F 101 NjHfN,NH2
H 0
composition comprises a compound having a structure of CI or
pharmaceutically acceptable salt thereof. In some cases, the composition
comprises a
0
H
0 NJYN,NH2
H
0
CI
compound having a structure F , or a
pharmaceutically acceptable
salt thereof.
[0017] Further provided are methods of inhibiting PAI-1 by contacting PAI-1
with a
compound as disclosed herein. Also provided are methods of treating a disorder
associated
with aberrant PAI-1 activity comprising administering to a subject in need
thereof a
compound as disclosed herein in an amount effective to treat the disorder. In
some cases,
the disorder is cancer, septicemia, obesity, insulin resistance, a disease or
disorder
associated with dysregulation of lipid metabolism, a disease or disorder
associated with an

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
elevated level of VLDL or LDL, high cholesterol, a proliferative disease or
disorder, fibrosis
and fibrotic disease, inflammatory bowel disease, coagulation homeostasis,
cerebrovascular
disease, microvascular disease, hypertension, dementia, atherosclerosis,
osteoporosis,
osteopenia, arthritis, asthma, heart failure, arrhythmia, angina, hormone
insufficiency,
Alzheimer's disease, hypertension, inflammation, sepsis, fibrinolytic
disorder, stroke,
dementia, coronary heart disease, myocardial infarction, stable and unstable
angina,
vascular disease, peripheral arterial disease, acute vascular syndrome,
thrombosis,
prothrombosis, deep vein thrombosis, pulmonary embolism, cerebrovascular
disease,
microvascular disease, hypertension, diabetes, hyperglycemia,
hyperinsulinemia, malignant
lesions, premalignant lesions, gastrointestinal malignancies, liposarcoma,
epithelial tumor,
and psoriasis, an extracellular matrix accumulation disorder, neoangiogenesis,
myelofibrosis, fibrinolytic impairment, polycystic ovary syndrome, bone loss
induced by
estrogen deficiency, angiogenesis, neoangiogenesis, myelofibrosis, or
fibrinolytic
impairment. In various cases, the disease or disorder involving thrombosis or
prothrombosis
is formation of atherosclerotic plaques, venous thrombosis, arterial
thrombosis, myocardial
ischemia, atrial fibrillation, deep vein thrombosis, a coagulation syndrome,
pulmonary
thrombosis, cerebral thrombosis, a thromboembolic complication of surgery, and
peripheral
arterial occlusion. In some cases, the disorder is fibrosis, and more
particularly, can be
pulmonary fibrosis, renal fibrosis, cardiac fibrosis, hepatic fibrosis, or
scleroderma. In some
cases, the disorder is inflammatory bowel disease, and more particularly, can
be Crohn's
disease or ulcerative colitis. In some cases, the extracellular matrix
accumulation disorder is
renal fibrosis, chronic obstructive pulmonary disease, polycystic ovary
syndrome, restenosis,
renovascular disease, diabetic nephropathy, or organ transplant rejection.
[0018] Further provided are methods of modulating cholesterol, lipid
clearance, and/or
lipid uptake in a subject with an elevated level of PAI-1 comprising
administering to the
subject an effective amount of a compound disclosed herein in an amount
effective to
decrease the elevated level of PAI and modulate cholesterol, lipid clearance,
and/or lipid
uptake in the subject. In some cases, the compound increases circulating high
density
lipoprotein (HDL) and/or decreases circulating very low density lipoprotein
(VLDL) in the
subject. In various cases, the compound inhibits apolipoprotein E (ApoE) or
apolipoprotein
A (ApoA) binding to VLDL-R. In various cases, the compound decreases HDL or
apolipoprotein E (ApoE) or apolipoprotein A (ApoA) binding to an ApoA
receptor. In various
cases, the compound decreases PAI-1 binding to apolipoprotein E (ApoE). In
various cases,
the compound decreases PAI-1 binding to apolipoprotein A (ApoA). In various
cases, the
compound decreases PAI-1 binding to VLDL. In various cases, the compound binds
to PAI-
1 in the presence of vitronectin. In various cases, the compound binds to PAI-
1 in the
presence of urokinase type plasminogen activator (uPA).
6

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0019] In any of the methods disclosed herein, the subject can be human.
[0020] The foregoing summary is not intended to define every aspect of the
invention, and
additional aspects are described in other sections, such as the Detailed
Description. The
entire document is intended to be related as a unified disclosure, and it
should be
understood that all combinations of features described herein are
contemplated, even if the
combination of features are not found together in the same sentence, or
paragraph, or
section of this document.
BRIEF DESCRIPTION OF THE FIGURES
[0021] Figure 1 shows the Serpin activity in the presence of various
concentrations of
CDE-517.
[0022] Figure 2 shows the Serpin activity in the presence of various
concentrations of
CDE-252.
[0023] Figure 3 shows the Serpin activity in the presence of various
concentrations of
CDE-519.
[0024] Figure 4 shows the Serpin activity in the presence of various
concentrations of
CDE-520.
[0025] Figure 5 shows the Serpin activity in the presence of various
concentrations of
CDE-264.
[0026] Figure 6 shows the Serpin activity in the presence of various
concentrations of
CDE-295.
[0027] Figure 7 shows the Serpin activity in the presence of various
concentrations of
CDE-234.
[0028] Figure 8 shows the Serpin activity in the presence of various
concentrations of
CDE-241.
[0029] Figure 9 shows the Serpin activity in the presence of various
concentrations of
CDE-246.
[0030] Figure 10 shows the Serpin activity in the presence of various
concentrations of
CDE-413.
[0031] Figure 11 shows the Serpin activity in the presence of various
concentrations of
CDE-415.
[0032] Figure 12 shows the Serpin activity in the presence of various
concentrations of
CDE-412.
7

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0033] Figure 13 shows the Serpin activity in the presence of various
concentrations of
CDE-248.
[0034] Figure 14 shows the Serpin activity in the presence of various
concentrations of
CDE-266.
[0035] Figure 15 shows the Serpin activity in the presence of various
concentrations of
CDE-301.
[0036] Figure 16 shows the Serpin activity in the presence of various
concentrations of
CDE-307.
[0037] Figure 17 shows the Serpin activity in the presence of various
concentrations of
CDE-340.
[0038] Figure 18 shows the Serpin activity in the presence of various
concentrations of
CDE-422.
[0039] Figure 19 shows the Serpin activity in the presence of various
concentrations of
CDE-423.
[0040] Figure 20 shows the Serpin activity in the presence of various
concentrations of
CDE-424.
[0041] Figure 21 shows the Serpin activity in the presence of various
concentrations of
CDE-446.
DETAILED DESCRIPTION
0
H
lei N)YN H2
H
0
X
[0042] Provided herein is a PAI-1
inhibitor having a structure: OCF3 ,
wherein X is Cl or F, or a pharmaceutically acceptable salt thereof. In some
cases, the
0 H
N,
(101 N)..r N H2
H
0
CI
compound has a structure OCF3 or a
pharmaceutically acceptable salt
0 H
N,
F lei N)..r N H2
H
0
thereof. In some cases, the compound has a structure OCF3 or a
pharmaceutically acceptable salt thereof. Further provided is a PAI-1
inhibitor having a
8

CA 03070303 2020-01-16
WO 2019/023526 PCT/US2018/043998
0
H
F0 N)..rN,NH2
H
0
structure CI or a pharmaceutically acceptable salt thereof.
Further
0
H
401 N)YN,NH2
H
0
CI
provided is a PAI-1 inhibitor having a structure F , or a
pharmaceutically acceptable salt thereof. Also provided are pharmaceutical
compositions
comprising one or more of these compounds or a salt thereof.
[0043] As used herein, the term "pharmaceutically acceptable salt" refers to
those salts
which are, within the scope of sound medical judgment, suitable for use in
contact with the
tissues of humans and lower animals without undue toxicity, irritation,
allergic response and
the like, and are commensurate with a reasonable benefit/risk ratio.
Pharmaceutically
acceptable salts are well known in the art. For example, S. M. Berge et al.
describe
pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences,
1977, 66, 1-19,
which is incorporated herein by reference. Pharmaceutically acceptable salts
of the
compounds of this disclosure include those derived from suitable inorganic and
organic
acids and bases. Examples of pharmaceutically acceptable, nontoxic acid
addition salts are
salts of an amino group formed with inorganic acids such as hydrochloric acid,
hydrobromic
acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids
such as acetic
acid, trifluoroacetic acid, oxalic acid, maleic acid, tartaric acid, citric
acid, succinic acid or
malonic acid or by using other methods used in the art such as ion exchange.
Other
pharmaceutically acceptable salts include adipate, alginate, ascorbate,
aspartate,
benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate,
camphorsulfonate,
citrate, cyclopentanepropionate, digluconate, dodecylsulf ate,
ethanesulfonate, formate,
fumarate, glucoheptonate, glycerophosphate, gluconate, glutamate, hemisulfate,

heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate,
lactate,
laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-
naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, pal mitate,
pamoate, pectinate,
persulf ate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate,
stearate, succinate,
sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate
salts, and the like.
Salts of compounds containing a carboxylic acid or other acidic functional
group can be
prepared by reacting with a suitable base. Such salts include, but are not
limited to, alkali
metal, alkaline earth metal, aluminum salts, ammonium, N(C1_4alky1)4 salts,
and salts of
organic bases such as trimethylamine, triethylamine, morpholine, pyridine,
piperidine,
picoline, dicyclohexylamine, N,N'-dibenzylethylenediamine, 2-
hydroxyethylamine, bis-(2-
9

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
hydroxyethyl)amine, tri-(2-hydroxyethyl)amine, procaine, dibenzylpiperidine,
dehydroabietylamine, N,N'-bisdehydroabietylamine, glucamine, N-
methylglucamine,
collidine, quinine, quinoline, and basic amino acids such as lysine and
arginine. This
disclosure also envisions the quaternization of any basic nitrogen-containing
groups of the
compounds disclosed herein. Water or oil-soluble or dispersible products may
be obtained
by such quaternization. Representative alkali or alkaline earth metal salts
include sodium,
lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically
acceptable
salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and
amine
cations formed using counterions such as halide, hydroxide, carboxylate,
sulfate, phosphate,
nitrate, lower alkyl sulfonate and aryl sulfonate.
Methods of Using PAI-1 Inhibitors
[0044] As mentioned herein above, it is contemplated that methods disclosed
herein
include treating a disease or disorder associated with elevated levels of PAI-
1 comprising
administering a PAI-1 inhibitor. In one aspect, the subject is a mammal. In
some cases, the
mammalian subject is human.
[0045] In some embodiments, provided herein are PAI-1 inhibitor compounds and
methods of using the compounds in the treatment of many diseases or disorders
associated
with PAI-1 activity. Such conditions, e.g., diseases or disorders, include,
but are not limited
to, dysregulation of lipid metabolism, obesity, diabetes, polycystic ovary
syndrome, bone
loss induced by estrogen deficiency, fibrosis and fibrotic disease,
inflammation, cell
migration and migration-driven proliferation of cells, and angiogenesis or
thrombosis. In
some aspects, such inhibitors are also contemplated to be useful for
modulation of
endogenous fibrinolysis, and in conjunction with pharmacologic thrombolysis.
In various
aspects, provided herein are PAI-1 inhibitor compounds and methods of using
the
compounds in the treatment of acute diseases associated with high PAI-1
levels, such as,
but not limited to, sepsis, myocardial infarction, and thrombosis, compared to
PAI-1 levels in
normal subjects known not to suffer from sepsis, myocardial infarction, or
thrombosis. In
some aspects, the PAI-1 inhibitor compounds disclosed herein are used in
methods for
treating diseases and disorders associated with high PAI-1 levels, such as,
but not limited to,
cancer, atherosclerosis, insulin resistance, type 2 diabetes, and fibrotic
diseases compared
to PAI-1 levels in normal subjects known not to suffer from these diseases or
disorders. In
various aspects, provided herein are PAI-1 inhibitor compounds for regulating
lipid
metabolism, including increasing circulating HDL and/or decreasing circulating
VLDL in a
subject.
[0046] In various aspects, a PAI-1 inhibitor is useful in the treatment of
any condition,
including a disease or disorder, wherein the lowering of PAI-1 levels will
provide benefits.

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
The PAI-1 inhibitor is useful alone, or in combination with other compounds,
which may act
as to promote the reduction of PAI-1 levels.
[0047] The PAI-1 inhibitor can be formulated into an appropriate preparation
and
administered to one or more sites within the subject in a therapeutically
effective amount. In
some embodiments, the PAI-1 inhibitor-based therapy is effected via continuous
or
intermittent intravenous administration. In various aspects, the PAI-1
inhibitor-based therapy
is effected via continuous or intermittent intramuscular or subcutaneous
administration. In
other aspects, the PAI inhibitor-based therapy is effected via oral or buccal
administration.
By "effective amount" what is meant is an amount of PAI-1 inhibitor compound
that is
sufficient to support an observable change in the level of one or more
biological activities of
PAI-1, plasminogen activator, HDL, LDL, or VLDL and/or an observable change in
an
indication for which the method of treatment is intended. The change may be
reduced level
of PAI-1 activity. In some aspects, the change is an increase in plasminogen
activator,
and/or HDL and/or a reduction in LDL and VLDL.
[0048] In various aspects, administration of the compositions is systemic
or local, and in
still other aspects comprises a single site injection of a therapeutically-
effective amount of
the PAI-1 inhibitor composition. Any route known to those of skill in the art
for the
administration of a therapeutic composition disclosed herein is contemplated
including, for
example, intravenous, intramuscular, subcutaneous, oral, or a catheter for
long-term
administration.
[0049] In some cases, it is contemplated that the therapeutic composition is
delivered to
the patient at multiple sites. The multiple administrations are rendered
simultaneously or are
administered over a period of several hours. It is likewise contemplated that
the therapeutic
composition is taken on a regular basis via oral administration. In certain
cases, it is
beneficial to provide a continuous flow of the therapeutic composition.
Additional therapy is
administered on a period basis, for example, daily, weekly, or monthly.
[0050] In addition to therapies based solely on the delivery of the PAI-1
inhibitor
composition, combination therapy is specifically contemplated. It is
contemplated that the
PAI-1 inhibitor composition therapy is used similarly in conjunction with
other agents
commonly used for the treatment of elevated levels of PAI-1, LDL and VLDL.
[0051] To achieve the appropriate therapeutic outcome, using the methods and
compositions disclosed herein, it is further contemplated that a composition
comprising a
PAI-1 inhibitor and at least one other therapeutic agent (second therapeutic
agent) is
administered to a subject in need thereof. Such therapeutic agents include
drugs used to
manage cardiovascular disease including, but not limited to, cholesterol
lowering drugs, such
as statins, anti-inflammatories, and ACE inhibitors. Such drugs also include
drugs targeting
11

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
neurological disorders including, but not limited to drugs for targeting
stroke, seizures, and
Alzheimer's Disease. In another aspect, the additional agents include, but are
not limited to,
drugs targeting diabetes. These are all disorders associated with elevated
levels of PAI-1
and, therefore, it is contemplated that combination therapy may be used with
PAI-1 inhibitors
and other known therapies.
[0052] The combination therapy compositions are provided in a combined amount
effective to produce the desired therapeutic outcome in the treatment of
increased levels of
PAI-1, VLDL, or LDL and/or make a detectable change in an indication as
described herein.
This process involves administering the PAI-1 inhibitor and the second
agent(s) or factor(s)
at the same time. Methods thus include administering a single composition or
pharmacological formulation that includes both agents, or administering two
distinct
compositions or formulations, at the same time, wherein one composition
includes the PAI-1
inhibitor therapeutic composition and the other includes the second
therapeutic agent.
[0053] Alternatively, the PAI-1 inhibitor treatment precedes or follows the
second
therapeutic agent treatment by intervals ranging from minutes to weeks. In
embodiments
where the second therapeutic agent and the PAI-1 inhibitor are administered
separately, one
generally ensures that a significant period of time did not expire between the
times of each
delivery, such that the second therapeutic agent and the PAI-1 inhibitor are
able to exert an
advantageously combined effect. In such instances, it is contemplated that one
administers
both modalities within about 12-24 hours of each other, or alternately, within
about 6-12
hours of each other, or alternately, with a delay time of only about 12 hours.
In some
situations, it is desirable to extend the time period for treatment
significantly, however, where
several days (2, 3, 4, 5, 6 or 7) to several weeks (1, 2, 3, 4, 5, 6, 7 or 8)
lapse between the
respective administrations.
[0054] Systemic delivery of PAI-1 inhibitors to patients is a very efficient
method for
delivering a therapeutically effective amount of the compound to counteract
the immediate
clinical manifestations of a disease or disorder. Alternatively, local
delivery of the PAI-1
inhibitor and/or the second therapeutic agent is appropriate in certain
circumstances. In a
certain embodiment, it is contemplated that the PAI-1 inhibitor is delivered
to a patient for an
extended period of time. It is further contemplated that the PAI-1 inhibitor
is taken
throughout a patient's lifetime to lower PAI-1, VLDL and/or LDL levels.
Pharmaceutical Compositions
[0055] As mentioned herein above, provided herein are methods using
pharmaceutical
compositions comprising effective amounts of PAI-1 inhibitor together with
pharmaceutically
acceptable excipient, such as diluents, preservatives, solubilizers,
emulsifiers, adjuvants
and/or carriers useful in PAI-1 inhibitor therapy. Such compositions include
diluents of
various buffer content (e.g., Tris-HCI, acetate, phosphate), pH and ionic
strength; additives
12

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80),
anti-oxidants
(e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., thimersol,
benzyl alcohol),
and bulking substances (e.g., lactose, mannitol); incorporation of the
material into particulate
preparations of polymeric compounds, such as polylactic acid, polyglycolic
acid, etc., or in
association with liposomes or micelles. Such compositions will influence the
physical state,
stability, rate of in vivo release, and rate of in vivo clearance of the PAI-1
inhibitor. See, e.g.,
Remington's Pharmaceutical Sciences, 18th Ed. (1990) Mack Publishing Co.,
Easton, PA,
pages 1435-1712, which are herein incorporated by reference.
[0056] Sterile liquid compositions include solutions, suspensions, emulsions,
syrups and
elixirs. The compounds disclosed herein may be dissolved or suspended in the
pharmaceutically acceptable carrier, such as sterile water, sterile organic
solvent or a
mixture of both. In one aspect, the liquid carrier is one suitable for
parental injection. Where
the compounds are sufficiently soluble they can be dissolved directly in
normal saline with or
without the use of suitable organic solvents, such as propylene glycol or
polyethylene glycol.
If desired, dispersions of the finely divided compounds can be made-up in
aqueous starch or
sodium carbmmethyl cellulose solution, or in a suitable oil, such as arachis
oil. Liquid
pharmaceutical compositions, which are sterile solutions or suspensions, can
be utilized by
intramuscular, intraperitoneal or subcutaneous injection. In many instances a
liquid
composition form may be used instead of the preferred solid oral method of
administration.
[0057] It is preferred to prepare unit dosage forms of the compounds for
standard
administration regimens. In this way, the composition can be subdivided
readily into smaller
doses at the physician's direction. For example, unit dosages may be made up
in packeted
powders, vials or ampoules and, in one aspect, in capsule or tablet form. The
active
compound present in these unit dosage forms of the composition may be present
in an
amount of from about one gram to about fifteen grams or more, for single or
multiple daily
administration, according to the particular need of the patient. The daily
dose of active
compound will vary depending upon the route of administration, the size, age
and sex of the
patient, the severity of the disease state, and the response to the therapy as
traced by blood
analysis and the patient's recovery rate.
[0058] The precise dosage to be employed depends upon several factors
including the
host, whether in veterinary medicine or human medicine, the nature and
severity of the
condition, e.g., disease or disorder, being treated, the mode of
administration and the
particular active substance employed. The compounds may be administered by any

conventional route, in particular enterally, and, in one aspect, orally in the
form of tablets or
capsules. Administered compounds can be in the free form or pharmaceutically
acceptable
salt form as appropriate, for use as a pharmaceutical, particularly for use in
the prophylactic
or curative treatment of atherosclerosis and sequelae (angina pectoris,
myocardial infarction,
13

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
arrhythmias, heart failure, kidney failure, stroke, peripheral arterial
occlusion, and related
disease states). These measures will slow the rate of progress of the disease
state and
assist the body in reversing the process direction in a natural manner.
[0059] PAI-1 inhibitors or derivatives thereof may be formulated for
injection, or oral,
nasal, pulmonary, topical, or other types of administration as one skilled in
the art will
recognize. The formulation may be liquid or may be solid, such as lyophilized,
for
reconstitution.
[0060] PAI-1 inhibitor or derivatives thereof are useful in the treatment
of any of the acute
or chronic diseases or disorders associated with increased levels of PAI-1,
LDL, or VLDL.
Conditions (e.g., diseases or disorders) alleviated or modulated by the
administration of PAI-
1 inhibitor, in some aspects, are those characterized by increased levels of
VLDL and LDL.
Such conditions may be induced as a course of therapy for other purposes, such
as
chemotherapy or radiation therapy. It is contemplated that such conditions may
result from
genetic inheritance or be the side effect of another condition or medication.
[0061] The phrase "pharmaceutically or pharmacologically acceptable" refers to
molecular
entities and compositions that do not produce adverse, allergic, or other
untoward reactions
when administered to an animal or a human. As used herein, "pharmaceutically
acceptable
carrier" includes any and all solvents, dispersion media, coatings,
antibacterial and
antifungal agents, isotonic and absorption delaying agents and the like. The
use of such
media and agents for pharmaceutically active substances is well-known in the
art. Except
insofar as any conventional media or agent is incompatible with the vectors or
cells, its use
in therapeutic compositions is contemplated. Supplementary active ingredients
also can be
incorporated into the compositions.
[0062] The active compositions used in the methods disclosed herein include
classic
pharmaceutical preparations. Administration of these compositions will be via
any common
route so long as the target tissue is available via that route. The
pharmaceutical
compositions may be introduced into the subject by any conventional method,
e.g., by
intravenous, intradermal, intramusclar, intramammary, intraperitoneal,
intrathecal,
retrobulbar, intrapulmonary (e.g., term release); by oral, sublingual, nasal,
anal, vaginal, or
transdermal delivery, or by surgical implantation at a particular site. The
treatment may
consist of a single dose or a plurality of doses over a period of time.
[0063] The active compounds may be prepared for administration as solutions of
free
base or pharmacologically acceptable salts in water suitably mixed with a
surfactant, such as
hydroxypropylcellulose. Dispersions also can be prepared in glycerol, liquid
polyethylene
glycols, and mixtures thereof and in oils. Under ordinary conditions of
storage and use,
these preparations contain a preservative to prevent the growth of
microorganisms.
14

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0064] The pharmaceutical forms suitable for injectable use include sterile
aqueous
solutions or dispersions and sterile powders for the extemporaneous
preparation of sterile
injectable solutions or dispersions. In all cases, the form must be sterile
and must be fluid to
the extent that easy syringability exists. It must be stable under the
conditions of
manufacture and storage and must be preserved against the contaminating action
of
microorganisms, such as bacteria and fungi. The carrier can be a solvent or
dispersion
medium containing, for example, water, ethanol, polyol (for example, glycerol,
propylene
glycol, and liquid polyethylene glycol, and the like), suitable mixtures
thereof, and vegetable
oils. The proper fluidity can be maintained, for example, by the use of a
coating, such as
lecithin, by the maintenance of the required particle size in the case of
dispersion and by the
use of surfactants. The prevention of the action of microorganisms can be
brought about by
various antibacterial and antifungal agents, for example, parabens,
chlorobutanol, phenol,
sorbic acid, thimerosal, and the like. In many cases, it will be preferable to
include isotonic
agents (for example, sugars or sodium chloride). Prolonged absorption of the
injectable
compositions can be brought about by the use in the compositions of agents
delaying
absorption (for example, aluminum monostearate and gelatin).
[0065] Sterile injectable solutions are prepared by incorporating the active
compounds in
the required amount in the appropriate solvent with several of the other
ingredients
enumerated above, as required, followed by filtered sterilization. Generally,
dispersions are
prepared by incorporating the various sterilized active ingredients into a
sterile vehicle that
contains the basic dispersion medium and the required other ingredients from
those
enumerated above. In the case of sterile powders for the preparation of
sterile injectable
solutions, the preferred methods of preparation are vacuum-drying and freeze-
drying
techniques that yield a powder of the active ingredient plus any additional
desired ingredient
from a previously sterile-filtered solution thereof.
[0066] As used herein, "pharmaceutically acceptable carrier" includes any and
all
solvents, dispersion media, coatings, antibacterial and antifungal agents,
isotonic and
absorption delaying agents and the like. The use of such media and agents for
pharmaceutical active substances is well-known in the art. Except insofar as
any
conventional media or agent is incompatible with the active ingredient, its
use in the
therapeutic compositions is contemplated. Supplementary active ingredients
also can be
incorporated into the compositions.
[0067] For oral administration of the compositions, a PAI-1 inhibitor may be
incorporated
with excipients and used in the form of non-ingestible mouthwashes and
dentifrices. A
mouthwash may be prepared incorporating the active ingredient in the required
amount in an
appropriate solvent, such as a sodium borate solution (Dobell's Solution).
Alternatively, the
active ingredient may be incorporated into an antiseptic wash containing
sodium borate,

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
glycerin and potassium bicarbonate. The active ingredient may also be
dispersed in
dentifrices, including: gels, pastes, powders and slurries. The active
ingredient may be
added in a therapeutically effective amount to a paste dentifrice that may
include water,
binders, abrasives, flavoring agents, foaming agents, and humectants.
[0068] The compositions used in the methods may be formulated in a neutral or
salt form.
Pharmaceutically-acceptable salts include the acid addition salts (formed with
the free amino
groups of the protein) and which are formed with inorganic acids such as, for
example,
hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic,
tartaric, mandelic,
and the like. Salts formed with the free carboxyl groups also can be derived
from inorganic
bases such as, for example, sodium, potassium, ammonium, calcium, or ferric
hydroxides,
and such organic bases as isopropylamine, trimethylamine, histidine, procaine
and the like.
[0069] The compositions used in the methods may be formulated in micelles or
liposomes.
Such formulations include sterically stabilized micelles or liposomes and
sterically stabilized
mixed micelles or liposomes. Such formulations can facilitate intracellular
delivery, since
lipid bilayers of liposomes and micelles are known to fuse with the plasma
membrane of
cells and deliver entrapped contents into the intracellular compartment.
[0070] Upon formulation, solutions will be administered in a manner compatible
with the
dosage formulation and in such amount as is therapeutically effective. The
formulations are
easily administered in a variety of dosage forms such as injectable solutions,
drug release
capsules and the like. For parenteral administration in an aqueous solution,
for example, the
solution should be suitably buffered if necessary and the liquid diluent first
rendered isotonic
with sufficient saline or glucose. These particular aqueous solutions are
especially suitable
for intravenous, intramuscular, subcutaneous and intraperitoneal
administration.
[0071] Generally, an effective amount of a PAI-1 inhibitor, or derivatives
thereof, will be
determined by the age, weight, and condition or severity of disease or
disorder of the
recipient. See, Remington's Pharmaceutical Sciences, supra, pages 697-773,
herein
incorporated by reference. Typically, a dosage of between about 0.001 pg/kg
body
weight/day to about 1000 pg/kg body weight/day, may be used, but more or less,
as a skilled
practitioner will recognize, may be used. Dosing may be one or more times
daily, or less
frequently, and may be in conjunction with other compositions as described
herein. It should
be noted that the disclosure is not limited to the dosages recited herein.
[0072] By initiating the treatment regimen with a minimal daily dose of about
one gram,
the blood levels of PAI-1 and the patient's symptomatic relief analysis may be
used to
determine whether a larger dose is indicated. One skilled in the art will
appreciate that the
appropriate dosage levels for treatment will thus vary depending, in part,
upon the molecule
delivered, the indication for which the PAI-1 inhibitor compound is being
used, the route of
16

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
administration, and the size (body weight, body surface or organ size) and
condition (the age
and general health) of the patient. Accordingly, the clinician may titer the
dosage and may
modify the route of administration to obtain the optimal therapeutic effect. A
typical dosage
may range from about 0.1 g/kg to up to about 100 mg/kg or more, depending on
the factors
mentioned above. In other embodiments, the dosage may range from 0.1 g/kg up
to about
100 mg/kg; or 1 g/kg up to about 100 mg/kg; or 5 g/kg up to about 100 mg/kg.
[0073] "Unit dose" is defined as a discrete amount of a therapeutic
composition dispersed
in a suitable carrier. Parenteral administration may be carried out with an
initial bolus
followed by continuous infusion to maintain therapeutic circulating levels of
drug product.
Those of ordinary skill in the art will readily optimize effective dosages and
administration
regimens as determined by good medical practice and the clinical condition of
the individual
patient.
[0074] The frequency of dosing will depend on the pharmacokinetic parameters
of the
agents and the routes of administration. The optimal pharmaceutical
formulation will be
determined by one of skill in the art depending on the route of administration
and the desired
dosage. See, for example, Remington's Pharmaceutical Sciences, supra, pages
1435-1712,
incorporated herein by reference. Such formulations may influence the physical
state,
stability, rate of in vivo release and rate of in vivo clearance of the
administered agents.
Depending on the route of administration, a suitable dose may be calculated
according to
body weight, body surface areas or organ size. Further refinement of the
calculations
necessary to determine the appropriate treatment dose is routinely made by
those of
ordinary skill in the art without undue experimentation, especially in light
of the dosage
information and assays disclosed herein, as well as the pharmacokinetic data
observed in
animals or human clinical trials.
[0075] Appropriate dosages may be ascertained through the use of established
assays for
determining level of myocardial infarct in conjunction with relevant dose-
response data. The
final dosage regimen will be determined by the attending physician,
considering factors that
modify the action of drugs, e.g., the drug's specific activity, severity of
the damage and the
responsiveness of the patient, the age, condition, body weight, sex and diet
of the patient,
the severity of any infection, time of administration and other clinical
factors. As studies are
conducted, further information will emerge regarding appropriate dosage levels
and duration
of treatment.
[0076] It will be appreciated that the pharmaceutical compositions and
treatment methods
disclosed herein are useful in fields of human medicine and veterinary
medicine. Thus the
subject to be treated is in one aspect a mammal. In another aspect, the mammal
is a
human.
17

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0077] In addition, further contemplated is a kit containing components
comprising a
composition comprising a PAI-1 inhibitor; and optionally, at least one
additional factor useful
in the treatment of the acute and chronic diseases and disorders discussed
herein.
Uses of Compounds in the Treatment of Diseases or Disorders
[0078] Provided herein is the use of compounds as disclosed for the production
of a
medicament for the treatment or prevention of any disease or disorder
discussed herein.
[0079] The compounds are inhibitors of the serine protease inhibitor PAI-1,
and are
therefore useful in the treatment or prophylaxis of those processes which
involve the
production and/or action of PAI-1. Thus, the compounds, in various aspects,
are useful in
preventing or reducing thrombosis, promoting thrombolysis, reducing fibrosis
regulating lipid
metabolism as described herein. In some aspects, the compounds are useful in
treating
high cholesterol and diseases or disorders associated with elevated levels of
PAI-1. In
various aspects, the compounds are useful in treating elevated levels of VLDL
or LDL. In
some aspects, the compounds are useful in elevating HDL.
[0080] In some aspects, provided are the uses of these inhibitors for the
treatment of a
disease or disorder associated with PAI-1 activity. Such diseases or disorders
include, but
are not limited to, inflammation, cell migration and migration-driven
proliferation of cells, and
angiogenesis or thrombosis. Such inhibitors are also contemplated to be useful
for
modulation of endogenous fibrinolysis, and in conjunction with pharmacologic
thrombolysis.
[0081] The compounds are useful in the treatment or prevention of insulin
resistance,
obesity, non-insulin dependent diabetes mellitus, cardiovascular disease,
thrombotic events
associated with coronary artery and cerebrovascular disease. The compounds are
also
useful for inhibiting the disease process involving the thrombotic and
prothrombotic states
which include, but are not limited to, formation of atherosclerotic plaques,
venous and
arterial thrombosis, myocardial ischemia, atrial fibrillation, deep vein
thrombosis, coagulation
syndromes, pulmonary thrombosis, cerebral thrombosis, thromboembolic
complications of
surgery (such as joint replacement), and peripheral arterial occlusion. These
compounds
are also useful in treating stroke associated with or resulting from atrial
fibrillation.
[0082] The compounds are also used in the treatment or prophylaxis of high
cholesterol
and diseases or disorders associated with such a condition.
[0083] The compounds may also be used in the treatment of diseases or
disorders
associated with extracellular matrix accumulation, including, but not limited
to, renal fibrosis,
chronic obstructive pulmonary disease, polycystic ovary syndrome, restenosis,
renovascular
disease and organ transplant rejection.
18

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0084] The compounds may also be used in the treatment of fibrosis, including,
but not
limited to, pulmonary fibrosis, renal fibrosis, cardiac fibrosis, hepatic
fibrosis, and
scleroderma.
[0085] The compounds may also be used in the treatment of inflammatory bowel
disease,
including, but not limited to, Crohn's disease and ulcerative colitis.
[0086] The compounds may also be used in the treatment of malignancies, and
diseases
or disorders associated with neoangiogenesis (such as diabetic retinopathy).
[0087] The compounds may also be used in conjunction with and following
processes or
procedures involving maintaining blood vessel patency, including vascular
surgery, vascular
graft and stent patency, organ, tissue and cell implantation and
transplantation.
[0088] The compounds may also be used in the treatment of Alzheimer's disease.
This
method may also be characterized as the inhibition of plasminogen activator by
PAI-1 in a
mammal, particularly a human, experiencing or subject to Alzheimer's disease.
This method
may also be characterized as a method of increasing or normalizing levels of
plasmin
concentration in a mammal, particularly those experiencing or subject to
Alzheimer's
disease.
[0089] The compounds may be used for the treatment of myelofibrosis with
myeloid
metaplasia by regulating stromal cell hyperplasia and increases in
extracellular matrix
proteins.
[0090] The compounds may also be used in conjunction with protease inhibitor-
containing
highly active antiretroviral therapy (HAART) for the treatment of diseases or
disorders which
originate from fibrinolytic impairment and hypercoagulability of HIV-1
infected patients
receiving such therapy.
[0091] The compounds may be used for the treatment of diabetic nephropathy and
renal
dialysis associated with nephropathy.
[0092] The compounds may be used to treat cancer, septicemia, proliferative
diseases,
such as psoriasis, improving coagulation homeostasis, cerebrovascular
diseases,
microvascular disease, hypertension, dementia, atherosclerosis, osteoporosis,
arthritis,
asthma, heart failure, arrhythmia, angina, and as a hormone replacement agent,
treating,
preventing or reversing progression of atherosclerosis, Alzheimer's disease,
osteoporosis,
osteopenia; reducing inflammatory markers, fibrinolytic disorder, reducing C-
reactive protein,
or preventing or treating low grade vascular inflammation, stroke, dementia,
coronary heart
disease, primary and secondary prevention of myocardial infarction, stable and
unstable
angina, primary prevention of coronary events, secondary prevention of
cardiovascular
events, peripheral vascular disease, peripheral arterial disease, acute
vascular syndromes,
19

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
deep vein thrombosis, pulmonary embolism, reducing the risk of undergoing a
myocardial
revascularization procedure, microvascular diseases such as nephropathy,
neuropathy,
retinopathy and nephrotic syndrome, hypertension, Type 1 and 2 diabetes and
related
diseases, obesity, insulin resistance, hyperglycemia, hyperinsulinemia,
malignant lesions,
premalignant lesions, gastrointestinal malignancies, liposarcomas and
epithelial tumors,
proliferative diseases such as psoriasis, improving coagulation homeostasis,
and/or
improving endothelial function, and all forms of cerebrovascular diseases.
[0093] The compounds disclosed herein can be used for the topical applications
in wound
healing for prevention of scarring.
[0094] The compounds disclosed herein can be used in the treatment of
inflammatory
diseases, septic shock and the vascular damage associated with infections and
for the
treatment of blood and blood products used in dialysis, blood storage in the
fluid phase,
especially ex vivo platelet aggregation. The compounds may also be used in
combination
with prothrombolytic, fibrinolytic and anticoagulant agents. The present
compounds may also
be added to human plasma during the analysis of blood chemistry in hospital
settings to
determine the fibrinolytic capacity thereof.
[0095] Further provided herein are methods for treating, preventing,
ameliorating or
inhibiting each of the maladies mentioned herein in a mammal, in one aspect,
in a human,
the method(s) each comprising administering to a mammal in need of such
treatment,
prevention, amelioration or inhibition a pharmaceutically or therapeutically
effective amount
of a compound disclosed herein, or a pharmaceutically acceptable salt thereof.
[0096] The compounds disclosed herein can also be used to treat cancer
including, but
not limited to, breast and ovarian cancer, and as imaging agents for the
identification of
metastatic cancers.
[0097] It will be understood that a pharmaceutically or therapeutically
effective amount of
a compound herein refers to an amount of the compound in question which will
sufficiently
inhibit the serine protease inhibitor PAI-1 in the mammal in need thereof to a
sufficient extent
to provide a desirable improvement in the condition in question or provide
sufficient inhibition
of the serine protease inhibitor PAI-1 to prevent, inhibit or limit the onset
of the physiological
basis for the malady or condition in question.
EXAMPLES
[0098] Synthesis of Compound CDE 517:
0
J..ro,
0 11 o
a
ocF,

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
[0099] Ethyl 2-((4-chloro-3-(trifluoromethoxy)benzyl)amino)-2-oxoacetate: A
stirring
solution of 4-chloro-3-(trifluoromethoxy)benzylamine (771.9 mg, 3.42 mmol) and
pyridine
(830 pL, 10.26 mmol) in dichloromethane (10 mL) was cooled in an ice bath.
Ethyl oxalyl
chloride (385 pL, 3.42 mmol) was added dropwise and the mixture was stirred
for 24 hours
at room temperature. The reaction mixture was diluted with ethyl acetate and
washed with
0.2N HCI (2x), saturated aqueous NaHCO3 (2x), and brine (1x). The organic
layer was dried
over magnesium sulfate, filtered, and concentrated in vacuo to afford 1.0726 g
(96% yield) of
product as a clear oil. 1H-NMR (DMSO-d6, 400MHz) 6 9.5 (t, J=6 Hz, 1H), 7.62
(d, J=8.2 Hz,
1H), 7.44 (bs, 1H), 7.3 (dd, J=8.7, 1.8 Hz, 1H), 4.33 (d, J=6.4 Hz, 2H), 4.21
(q, J=6.9 Hz,
2H), 1.23 (t, J=6.9 Hz, 3H).
o
)yNFINH2
*I 11 o
a
ocF,
[00100] N-(4-chloro-3-(trifluoromethoxy)benzy1)-2-hydraziny1-2-oxoacetamide
(CDE-
517): To a solution of ethyl 2-((4-chloro-3-(trifluoromethoxy)benzyl)amino)-2-
oxoacetate
(1.0726 g, 3.29 mmol) in absolute ethanol (30 mL) was added 50% hydrazine
hydrate (425
pL, 6.59 mmol) dropwise and stirred for 2 hours.. The solid was filtered,
dried in vacuo, and
then triturated with boiling deionized water to afford 0.7191 g (70.2% yield)
of product as a
white solid. 1H-NMR (DMSO-d6, 400 MHz) 510.1 (bs, 1H), 9.37 (t, J=6.4 Hz, 1H),
7.61 (d,
J=8.2 Hz, 1H), 7.42 (bs, 1H), 7.28 (dd, J=8.3, 1.8 Hz, 1H), 4.5 (d, J=3.6 Hz,
2H), 4.31 (d,
J=6.4 Hz, 2H); 13C-NMR (DMSO-d6, 100 MHz) 5160.6, 158.3, 144.3, 141.1, 131.4,
128.5,
124.8, 122.4, 120.6 (q, J=256.5 Hz), 41.8.
[00101] Synthesis of Compound CDE-415:
[00102] N-(3-chloro-4-fluorobenzy1)-2-hydraziny1-2-oxoacetamide (CDE-415): To
a
solution of 3-chloro-4-fluorobenzylamine (230 pL, 1.83 mmol) and pyridine (296
pL, 3.66
mmol) in dichloromethane (5 ml), ethyl 2-chloro-2-oxoacetate (215 LL, 1.92
mmol) was
added dropwise over an ice bath. The solution was removed from the ice bath
after 5
minutes and left to warm to room temperature. The reaction mixture was diluted
with ethyl
acetate, washed with 0.2 N HCI (2x) and saturated NaHCO3 (1x), dried with
MgSO4, filtered
and concentrated in vacuo to give 0.336 g of ethyl 2-(3-chloro-4-
fluorobenzylamino)-2-
oxoacetate as a white solid (71% yield). 1H NMR (CDCI3, 400 MHz) 8 7.42 (s,
1H), 7.34 (dd,
J=2.3, 6.9 Hz, 1H), 7.17 (M, 1H), 7.10 (t, J=8.7 Hz, 1H), 4.46 (d, J=6.0 Hz,
2H), 4.35 (q,
J=7.3 Hz, 2H), 1.39 (t, J=6.9 Hz, 3H). To a solution of ethyl 2-(3-chloro-4-
fluorobenzylamino)-2-oxoacetate (211.9 mg, 0.816 mmol) in ethanol (6 ml), 50%
hydrazine
hydrate (102 pL) was added dropwise. The reaction was stirred overnight at
room
temperature. The product was filtered from the mixture and dried in vacuo,
providing 0.188 g
21

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
of N-(3-chloro-4-fluorobenzy1)-2-hydraziny1-2-oxoacetamide as a white solid
(94% yield). 1H
NMR (DMSO-d6, 400 MHz) 8 10.01 (s, 1H), 9.28 (t, J=6.4 Hz, 1H), 7.42 (dd,
J=1.8, 5.5 Hz,
1H), 7.32 (t, J=8.7 Hz, 1H), 7.23 (m, 1H), 4.48 (s, 2H), 4.25 (d, J=6.4 Hz,
2H); 13C NMR
(DMSO-d6, 100 MHz) 8 160.48, 158.43, 156.75 (d, J=244 Hz), 137.32, 129.97,
128.63,
119.62 (J=18 Hz), 117.25 (J=20 Hz), 41.66.
[00103] Synthesis of Compound CDE-412:
[00104] N-(4-chloro-3-fluorobenzy1)-2-hydraziny1-2-oxoacetamide (CDE-412): To
a
solution of 4-chloro-3-fluorobenzylamine (225 1_, 1.83 mmol) and pyridine
(296 1_, 3.66
mmol) in dichloromethane (5 ml), ethyl 2-chloro-2-oxoacetate (215 LL, 1.92
mmol) was
added dropwise over an ice bath. The solution was removed from the ice bath
after 10
minutes and left to warm to room temperature. The reaction mixture was diluted
with ethyl
acetate, washed with 0.2 N HCI (2x) and saturated NaHCO3 (1x), dried with
MgSO4, filtered
and concentrated in vacuo to give 0.3878 g (82% yield) of ethyl 2-(4-chloro-3-
fluorobenzylamino)-2-oxoacetate as a white solid. 1H NMR (CDCI3, 400 MHz) 8
7.45 (s, 1H),
7.36 (t, J=7.96 Hz, 1H), 7.09 (d, J=9.6 Hz, 1H), 7.02 (d, J=8.3 Hz, 1H), 4.48
(d, J=6.4 Hz,
2H), 4.35 (q, J=6.9 Hz, 2H), 1.38 (t, J=6.9 Hz, 3H). To a solution of ethyl 2-
(4-chloro-3-
fluorobenzylamino)-2-oxoacetate (119.7 mg, 0.461 mmol) in ethanol (6 ml), 50%
hydrazine
hydrate (57 L) was added dropwise. The reaction was stirred overnight at room

temperature. The product was filtered from the mixture and dried in vacuo,
providing 59.0
mg of N-(4-chloro-3-fluorobenzy1)-2-hydraziny1-2-oxoacetamide as a white solid
(52% yield).
1H NMR (DMSO-d6, 400 MHz) 8 10.02 (s, 1H), 9.30 (t, J=6.4 Hz, 1H), 7.49 (t,
J=8.24 Hz,
1H), 7.24 (dd, J=1.8, 10.5 Hz, 1H), 7.08 (dd, J=1.36, 8.24Hz, 1H), 4.51 (s,
2H), 4.27 (d,
J=6.4 Hz, 2H); 13C NMR (DMSO-d6, 100 MHz) 8 160.54, 158.74, 158.40, 156.29,
141.29,
141.23, 130.99, 125.08, 125.05, 118.36, 118.19, 116.31, 116.10, 41.86.
[00105] Fluorometric IC50 plate assay for PAI-1 inhibitors: For assaying
PAI-1 inhibitor
activity in plasma, recombinant active human PAI-1 (Molecular Innovations) was
added to
PAI-1 depleted human plasma (Molecular Innovations) containing 10 g/mL
aprotinin
(Roche) to a concentration 20nM. Then 104 of this human plasma (with or
without PAI-1)
was added to wells containing 80 Lof buffer with increasing concentrations of
the PAI-1
inhibitor (Buffer: 40mM HEPES, 100mM NaCI, 0.005% Tween-20, pH 7.4, and 10%
DMSO)
and incubated for 15min at 23 C. Next, 104 of 25nM UPA (rheotromb) (final 2.5
nM) was
added to each reaction well and incubated for an additional 30 min at 24 C,
the final PAI-1
concentration was 2nM and the final uPA concentration was 2.5 nM. Following
this
incubation 1004 of buffer containing 100mM of the uPA fluorgenic substrate Z-
Gly-Gly-Arg-
AMC (Calbiochem) is added for a final concentration of 50 M, and the residual
uPA activity
in each reaction mixture was determined from the rate of AMC release by uPA
measured
22

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
with an excitation wavelength of 370nm and an emission wavelength of 440nm for
10
minutes at 23 C. Data are expressed as the residual PAI-1 activity as a
percent of the
control PAI-1 activity.
[00106] For assays in buffer or buffer containing 1.5% bovine serum albumin
(BSA) the
assay was as above except that no plasma or aprotinin was added and activity
was
determined in either 100mM NaCI, 40mM HEPES, 0.005% Tween-20, 10% DMSO, pH
7.4,
or in the same buffer containing 1.5% BSA. Results are shown in the figures.
Figure 1 is for
0
irki- Pi p '1:'
..,
CV '17
a compound of structure Qi31,-.3 (CDE-517). Figure 2 is for a compound
of
cx:f3 o i.4
f 'kr Pi 1 NH
0
structure \No (CDE-252). Figure 3 is for a compound of structure
Ck H3 9
.---)k..,- ---- N '=-= , . ?4 µNi-tz
k)r) " 8
(CDE-519). Figure 4 is for a compound of structure
MU 0 P
. 1,
).'''''' µ N II' 'NN2
L i H g
1
acf,3 (CDE-520). Figure 5 is for a compound of structure
7 N
==,. ...,,N, N
\\T=-=.;,',$ 0
F (CDE-264). Figure 6 is for a compound of structure
1.
..-- N._ ...-= ,...N,- == =
q "?'
.....-w*
T
OCHa (CDE-295). Figure 7 is for a compound of structure
0 H
)s, N .., ....--,,11,.. .....
1 ......, t...
(CDE-234). Figure 8 is for a compound of structure
0
..õ......,..,,,,,,,....: ,N..z.:
4
0 (CDE-241). Figure 9 is for a compound of structure
23

CA 03070303 2020-01-16
WO 2019/023526 PCT/US2018/043998
KIM = 0
r ti
N3M's k'k=-''''''N=='s"\qi'A, \ 1N1H2
C (CDE-246). Figure 10 is for a compound of structure
ti
0
¨ c:F4
(CDE-413).
0
N,
40/ IF\N NH2
0
[00107] Figure 11 is for a compound of structure CI (CDE-415).
0
N,
CI 101 N)Y NH2
" 0
Figure 12 is for a compound of structure F (CDE-
412). These
data compare to those of monohalophenyl compounds CDE-248 and CDE-266 (Figures
13
and 14, respectively), as well as to various other dihalophenyl compounds:
i1 U
0 ,
N NH,
II A - NH
!i
CV 0
(CDE-301, Figure 15);
= = 6
(CDE-307, Figure 16); (CDE-340, Figure 17);
0
!!
(CDE-422, Figure 18);
0
'1 4 ==it
,
(CDE-423, Figure 19);
24

CA 03070303 2020-01-16
WO 2019/023526
PCT/US2018/043998
0
=
n
N rsi4k,
g
(CDE-424, Figure 20); and
F 0
N
"m2
(CDE-446, Figure 21).

Representative Drawing

Sorry, the representative drawing for patent document number 3070303 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2018-07-27
(87) PCT Publication Date 2019-01-31
(85) National Entry 2020-01-16
Examination Requested 2023-07-25

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $210.51 was received on 2023-07-14


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-07-29 $100.00
Next Payment if standard fee 2024-07-29 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Registration of a document - section 124 2020-01-16 $100.00 2020-01-16
Registration of a document - section 124 2020-01-16 $100.00 2020-01-16
Application Fee 2020-01-16 $400.00 2020-01-16
Maintenance Fee - Application - New Act 2 2020-07-27 $100.00 2020-07-14
Maintenance Fee - Application - New Act 3 2021-07-27 $100.00 2021-07-08
Maintenance Fee - Application - New Act 4 2022-07-27 $100.00 2022-06-29
Maintenance Fee - Application - New Act 5 2023-07-27 $210.51 2023-07-14
Request for Examination 2023-07-27 $816.00 2023-07-25
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE REGENTS OF THE UNIVERSITY OF MICHIGAN
EASTERN MICHIGAN UNIVERSITY
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2020-01-16 1 57
Claims 2020-01-16 3 103
Drawings 2020-01-16 9 238
Description 2020-01-16 25 1,249
International Search Report 2020-01-16 2 86
National Entry Request 2020-01-16 10 366
Cover Page 2020-03-05 1 30
Request for Examination / Amendment 2023-07-25 9 257
Claims 2023-07-25 3 152